Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
PLoS One ; 14(12): e0226154, 2019.
Article En | MEDLINE | ID: mdl-31805144

Amiselimod (MT-1303) is a novel sphingosine 1-phosphate receptor-1 (S1P1 receptor) modulator with a more favorable cardiac safety profile than other S1P1 receptor modulators. MT-1303 phosphate (MT-1303-P), an active metabolite of MT-1303, exhibits S1P1 receptor agonism at a lower EC50 value than other S1P1 receptor modulators currently being developed. We aimed to evaluate the efficacy of MT-1303 and its mode of action in chronic colitis using an inflammatory bowel disease (IBD) model. Oral administration of MT-1303 (0.3 mg/kg) once daily for 3 days to mice almost completely abolished S1P1 receptor expression on CD4+ T cells from mesenteric lymph nodes, which corresponded to a marked decrease in CD4+ T cell count in peripheral blood, indicating that MT-1303-P acts as a functional antagonist of the S1P1 receptor. The potential benefit of MT-1303 for IBD was assessed using immunodeficient SCID mice with chronic colitis induced by adoptive transfer of CD4+CD45RBhigh T cells from BALB/c mice. An oral dose of 0.1 and 0.3 mg/kg MT-1303 administered daily one week after the cell transfer inhibited the development of chronic colitis with an efficacy comparable to that of an anti-mTNF-α mAb (250 µg/mouse). In addition, MT-1303 administration significantly reduced the number of infiltrating Th1 and Th17 cells into the lamina propria of the colon in colitis mice. Our results suggest that MT-1303 acts as a functional antagonist of the S1P1 receptor on lymphocytes, regulates lymphocyte trafficking, and inhibits infiltration of colitogenic Th1 and Th17 cells into the colon to inhibit the development of chronic colitis.


CD4-Positive T-Lymphocytes/transplantation , Colitis/drug therapy , Leukocyte Common Antigens/metabolism , Propanolamines/administration & dosage , Administration, Oral , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Colitis/immunology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Propanolamines/pharmacology , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors
2.
J Immunol Res ; 2019: 5821589, 2019.
Article En | MEDLINE | ID: mdl-31930150

Amiselimod (MT-1303) is a novel and selective sphingosine 1-phosphate receptor-1 (S1P1) modulator with a more favorable cardiac safety profile than other S1P1 receptor modulators. In this study, we evaluated the effects of MT-1303 on the progression of lupus nephritis in two well-known murine systemic lupus erythematosus (SLE) models, MRL/lpr and NZBWF1 mice, compared with those of FK506. Daily oral doses of 0.1 and 0.3 mg/kg MT-1303 not only inhibited the development of lupus nephritis when administered before onset in MRL/lpr and NZBWF1 mice but also improved symptoms of lupus nephritis when administered after onset in MRL/lpr mice. Its efficacy in these models was more potent or comparable to that of FK506 (1 and 3 mg/kg). In histological analysis, treatment with MT-1303 inhibited infiltration of T cells into the kidneys, mesangial expansion, and glomerular sclerosis. MT-1303 treatment resulted in a marked reduction in T cells and B cells in the peripheral blood and significantly inhibited increases in the number of plasma cells in the spleen and T cells in the kidneys. In addition, administration of MT-1303 suppressed elevations in serum anti-dsDNA antibody levels in MRL/lpr mice, but not in NZBWF1 mice. Our findings show that MT-1303 exhibits marked therapeutic effects on lupus nephritis in two SLE models, likely by reducing the infiltration of autoreactive T cells into the kidneys. These results suggest that MT-1303 has the potential to be used as a therapeutic agent for patients suffering from SLE, including lupus nephritis.


Kidney/drug effects , Lupus Nephritis/drug therapy , Propanolamines/therapeutic use , Sphingosine-1-Phosphate Receptors/drug effects , Animals , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Disease Models, Animal , Disease Progression , Female , Kidney/cytology , Kidney/immunology , Kidney/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Male , Mesangial Cells/drug effects , Mice , Mice, Inbred MRL lpr , Plasma Cells/drug effects , Plasma Cells/immunology , Sphingosine-1-Phosphate Receptors/metabolism , Spleen/cytology , Spleen/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Bioorg Med Chem Lett ; 26(5): 1365-70, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26869194

A phenotypic screening of thienodiazepines derived from a hit compound found through a binding assay targeting co-stimulatory molecules on T cells and antigen presenting cells successfully led to the discovery of a thienotriazolodiazepine compound (7f) possessing potent immunosuppressive activity. A chemical biology approach has succeeded in revealing that 7f is a first inhibitor of epigenetic bromodomain-containing proteins. 7f is expected to become an anti-cancer agent as well as an immunosuppressive agent.


Antineoplastic Agents/pharmacology , Azepines/pharmacology , CD28 Antigens/metabolism , Drug Discovery , Immunosuppressive Agents/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Azepines/chemical synthesis , Azepines/chemistry , CD28 Antigens/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Histone Acetyltransferases , Histone Chaperones , Humans , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/chemistry , Molecular Structure , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phenotype , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Article Ja | MEDLINE | ID: mdl-26016635

Mesenchymal stem cells (MSCs) possess multipotent capacity and exhibit immunoregulatory properties. In particular, MSCs can be easily isolated from various organs, can differentiate into various types of cells and generate regulatory T cells. Using human MSC derived from bone marrow and adipose tissue, we have clarified the following novel findings in vitro. 1) MSCs differentiated into osteoblasts or osteocytes under osteoblast-conditioned medium including the inflammatory stimuli such as IL-1. 2) The combination of IL-6 and soluble IL-6 receptor induced differentiation of MSCs to chondrocyte, whereas IL-17 inhibited their differentiation. 3) MSCs highly produced osteoprotegerin and inhibited osteoclastogenesis. Furthermore, we developed a local delivery system of MSCs by using nano-fiber scaffold. MSCs seeded on nano-fiber scaffold suppressed arthritis and bone destruction due to inhibition of systemic inflammatory reaction and immune response by suppressing T cell proliferation and reducing anti-type II collagen antibody production in vivo. Thus, our data may serve as a new strategy for MSC-based therapy in inflammatory diseases and an alternative delivery method for the treatment of destruction of bone and joints.


Arthritis, Rheumatoid/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Adipose Tissue/cytology , Antibody Formation , Bone Marrow Cells/cytology , Cell Differentiation , Collagen Type II/immunology , Culture Media , Humans , Interleukin-1 , Interleukin-17 , Interleukin-2 , Interleukin-6 , Lymphocyte Activation , Mesenchymal Stem Cells/metabolism , Nanofibers , Osteoblasts , Osteocytes , Osteoprotegerin/biosynthesis , Receptors, Interleukin-6 , T-Lymphocytes/immunology , Tissue Scaffolds
5.
Rheumatology (Oxford) ; 53(7): 1282-90, 2014 Jul.
Article En | MEDLINE | ID: mdl-24599911

OBJECTIVE: The mechanisms of ectopic calcification in inflammatory diseases are poorly understood. We investigated the effects of inflammatory cytokines on the mechanisms of calcification in human adipose tissue-derived mesenchymal stem cells (hADSCs). METHODS: The effects of inflammatory cytokines were evaluated using hADSCs cultured in osteoblast induction medium. mRNA expression was measured by real-time PCR and protein levels were measured by western blotting. Cell mineralization was evaluated by Alizarin Red S staining. RESULTS: In hADSCs, administration of IL-6/soluble IL-6 receptor (sIL-6R), TNF or IL-1ß accelerated calcification through enhanced expression of an osteoblast differentiation marker, runt-related transcription factor 2 (RUNX2). IL-6/sIL-6R had the greatest effect. The transcription of mRNA for receptor tyrosine kinase-like orphan receptor 2 (ROR2), involved in the non-canonical wingless-type (WNT) MMTV integration site pathway, was increased, while ß-catenin expression, an essential factor in the canonical WNT signalling pathway for osteoblast differentiation, did not change. Suppression of signal transducer and activator of transcription 3 (STAT3), but not STAT1, by small interfering RNA (siRNA) exerted a strong inhibitory effect on RUNX2 and ROR2 expression, and inhibited accelerated calcification. CONCLUSION: IL-6/sIL-6R stimulation accelerated the ROR2/WNT5A pathway in hADSCs in a STAT3-dependent manner, resulting in augmented calcification. These results suggest that the mechanisms of ectopic calcification accelerated by IL-6 in hADSCs may be involved in chronic inflammatory tissues and that IL-6 inhibitors may be beneficial in the treatment of ectopic calcification in inflammatory diseases.


Adipose Tissue/metabolism , Calcification, Physiologic/drug effects , Interleukin-6/pharmacology , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , STAT3 Transcription Factor/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Calcification, Physiologic/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , In Vitro Techniques , Interleukin-1beta/pharmacology , Interleukin-6/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology , Wnt Proteins/metabolism , Wnt-5a Protein , beta Catenin/metabolism
6.
Pharmaceuticals (Basel) ; 7(1): 1-17, 2013 Dec 23.
Article En | MEDLINE | ID: mdl-24366113

Interleukin (IL)-15 and IL-17 are thought to play an important role in the pathogenesis of rheumatoid arthritis (RA) because both pro-inflammatory cytokines are found in synovial fluid of RA patients. In this study, we examined the pharmacological profiles of Y-320, a new phenylpyrazoleanilide immunomodulator. Y-320 inhibited IL-17 production by CD4 T cells stimulated with IL-15 with IC50 values of 20 to 60 nM. Oral administration of Y-320 (0.3 to 3 mg/kg) significantly inhibited the development and progression of arthritis and joint destruction with reduction of IL-17 mRNA expression in arthritic joints of type II collagen-induced arthritis (CIA) in DBA/1J mice. Y-320 in combination with anti-murine tumor necrosis factor-α monoclonal antibody showed a synergistic effect on mouse CIA. Moreover, therapeutic treatment with Y-320 (0.3 and 1 mg/kg orally) ameliorated CIA in cynomolgus monkeys. Our results suggest that Y-320, an orally active inhibitor for IL-17 production, provides a useful therapy for RA.

7.
PLoS One ; 8(11): e79463, 2013.
Article En | MEDLINE | ID: mdl-24260226

OBJECTIVE: Mesenchymal stem cells (MSCs) can differentiate into cells of mesenchymal lineages, such as osteoblasts and chondrocytes. Here we investigated the effects of IL-17, a key cytokine in chronic inflammation, on chondrogenic differentiation of human MSCs. METHODS: Human bone marrow MSCs were pellet cultured in chondrogenic induction medium containing TGF-ß3. Chondrogenic differentiation was detected by cartilage matrix accumulation and chondrogenic marker gene expression. RESULTS: Over-expression of cartilage matrix and chondrogenic marker genes was noted in chondrogenic cultures, but was inhibited by IL-17 in a dose-dependent manner. Expression and phosphorylation of SOX9, the master transcription factor for chondrogenesis, were induced within 2 days and phosphorylated SOX9 was stably maintained until day 21. IL-17 did not alter total SOX9 expression, but significantly suppressed SOX9 phosphorylation in a dose-dependent manner. At day 7, IL-17 also suppressed the activity of cAMP-dependent protein kinase A (PKA), which is known to phosphorylate SOX9. H89, a selective PKA inhibitor, also suppressed SOX9 phosphorylation, expression of chondrogenic markers and cartilage matrix, and also decreased chondrogenesis. CONCLUSIONS: IL-17 inhibited chondrogenesis of human MSCs through the suppression of PKA activity and SOX9 phosphorylation. These results suggest that chondrogenic differentiation of MSCs can be inhibited by a mechanism triggered by IL-17 under chronic inflammation.


Chondrogenesis/drug effects , Interleukin-17/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Humans , Immunohistochemistry , Microscopy, Fluorescence , Phosphorylation/drug effects , SOX9 Transcription Factor/metabolism
8.
J UOEH ; 35(1): 33-7, 2013 Mar 01.
Article Ja | MEDLINE | ID: mdl-23475022

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovitis and bone destruction leading to irreversible joint deformity. The development of a novel treatment for RA aiming at joint repair is necessary. Recently, mesenchymal stem cells (MSCs) have been widely studied as a new therapeutic tool for the treatment of RA, due to their multipotency and also their immunosuppressive properties. We show here that MSCs inhibit osteoclast differentiation depending on the constitutive production of osteoprotegerin, a decoy receptor of receptor activator of nuclear factor kappa-B ligand (RANKL). Our results further indicate that MSCs are useful in RA treatment by preventing the progression of bone damage by inhibiting osteoclast differentiation. In addition, MSCs are likely to play important roles in bone metabolism and maintenance of joint formation. In conclusion, MSC is a promising tool for both anti-inflammation and bone repair for RA patients.


Arthritis, Rheumatoid/therapy , Mesenchymal Stem Cells/physiology , Osteoclasts/physiology , Humans , Osteoprotegerin/biosynthesis
9.
Arthritis Rheum ; 64(10): 3355-63, 2012 Oct.
Article En | MEDLINE | ID: mdl-22674197

OBJECTIVE: Mesenchymal stem cells (MSCs) are considered to be a novel tool for the treatment of rheumatoid arthritis (RA) because of their multipotency to differentiate into osteoblasts and chondrocytes, their immunosuppressive effects, and availability. The aim of this study was to assess the mechanisms of human MSC differentiation into osteoblasts under inflammatory conditions. METHODS: Human MSCs were cultured in commercialized osteogenic induction medium with inflammatory cytokines for up to 10 days. Osteoblast differentiation was detected by alkaline phosphatase staining and messenger RNA (mRNA) expression of multiple osteoblast markers. Mineralization was assessed by alizarin red S staining. RESULTS: Among the various cytokines tested, interleukin-1ß (IL-1ß) induced differentiation of human MSCs into osteoblasts, which was confirmed by alkaline phosphatase activity, expression of RUNX2 mRNA, and strong alizarin red S staining. Among various molecules of the Wnt family, Wnt-5a and receptor tyrosine kinase-like orphan receptor 2 (Ror2), a major receptor of Wnt-5a, were significantly induced in human MSCs by IL-1ß. Silencing of either WNT5A or ROR2 by small interfering RNA with 2 different sequences reduced alkaline phosphatase activity, RUNX2 expression, and alizarin red S staining of human MSCs induced by IL-1ß. CONCLUSION: IL-1ß effectively and rapidly induced human MSC differentiation into osteoblasts and mineralization, mainly through the noncanonical Wnt-5a/Ror2 pathway. These results suggest potential benefits of IL-1ß-treated human MSCs in the treatment of damaged bone as well as in the induction of self-renewal and self-repair of damaged tissue, including osseous tissue.


Cell Differentiation/physiology , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Proto-Oncogene Proteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Cell Differentiation/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteogenesis/physiology , Proto-Oncogene Proteins/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects , Wnt-5a Protein
10.
Arthritis Rheum ; 63(6): 1658-67, 2011 Jun.
Article En | MEDLINE | ID: mdl-21360521

OBJECTIVE: Mesenchymal stem cells (MSCs) have been proposed to be a useful tool for treatment of rheumatoid arthritis (RA), not only because of their multipotency but also because of their immunosuppressive effect on lymphocytes, dendritic cells, and other proinflammatory cells. Since bone destruction caused by activated osteoclasts occurs in RA, we undertook the present study to investigate the effect of MSCs on osteoclast function and differentiation in order to evaluate their potential use in RA therapy. METHODS: Human MSCs and peripheral blood mononuclear cells were cultured under cell-cell contact-free conditions with osteoclast induction medium. Differentiation into osteoclast-like cells was determined by tartrate-resistant acid phosphatase staining and expression of osteoclast differentiation markers. RESULTS: The number of osteoclast-like cells was decreased and expression of cathepsin K and nuclear factor of activated T cells c1 (NF-ATc1) was down-regulated by the addition of either MSCs or a conditioned medium obtained from MSCs. Osteoprotegerin (OPG) was constitutively produced by MSCs and inhibited osteoclastogenesis. However, osteoclast differentiation was not fully recovered upon treatment with either anti-OPG antibody or OPG small interfering RNA, suggesting that OPG had only a partial role in the inhibitory effect of MSCs. Moreover, bone-resorbing activity of osteoclast-like cells was partially recovered by addition of anti-OPG antibody into the conditioned medium. CONCLUSION: The present results indicate that human MSCs constitutively produce OPG, resulting in inhibition of osteoclastogenesis and expression of NF-ATc1 and cathepsin K in the absence of cell-cell contact. Therefore, we conclude that human MSCs exert a suppressive effect on osteoclastogenesis, which may be beneficial in inhibition of joint damage in RA.


Mesenchymal Stem Cells/metabolism , Osteoclasts/metabolism , Osteoprotegerin/biosynthesis , Acid Phosphatase/metabolism , Cathepsin K/biosynthesis , Cell Differentiation/drug effects , Cell Line , Coculture Techniques , Humans , Isoenzymes/metabolism , Mesenchymal Stem Cells/cytology , Monocytes , NFATC Transcription Factors/biosynthesis , Osteoclasts/cytology , Osteoclasts/drug effects , RNA, Small Interfering/pharmacology , Rheumatic Fever/therapy , Tartrate-Resistant Acid Phosphatase
11.
Inflamm Res ; 59(8): 659-66, 2010 Aug.
Article En | MEDLINE | ID: mdl-20217457

OBJECTIVES: The goal of this paper is to investigate the effects of activated complement C5a on vascular endothelium during vessel formation. METHODS: A human microvascular endothelial cell line (HMEC-1) derived from post-capillary venules in skin was used to measure DNA synthesis, proliferation and cell-cycle progression. In vitro ring-shaped formation by the cells was assessed by using type I collagen gel matrix and a cell-migration assay using the Chemotaxicell chamber. A Matrigel plug assay was performed to confirm the effect of C5a in vivo. RESULTS: C5a progressed the cell cycle of HMEC-1 into G2/M phases, and induced DNA synthesis and proliferation in a dose-dependent manner. C5a efficiently induced migration and ring-shaped structure formation both in vitro and in vivo. Furthermore, a C5a receptor antagonist (W-54011) suppressed all HMEC-1 activities including proliferation and migration. CONCLUSIONS: Proliferation, migration, and ring-shaped formation by HMEC-1 cells was induced by C5a. The actions were efficiently inhibited by a specific antagonist against C5a. Our results implicated C5a in vessel formation and as a potent target for management of inflammatory diseases.


Cell Movement/drug effects , Cell Proliferation/drug effects , Complement C5a/immunology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Neovascularization, Pathologic , Animals , Cell Cycle/drug effects , Cell Line , Complement Activation , Complement C5a/antagonists & inhibitors , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans
...